Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
2.
SLAS Technol ; 23(3): 243-251, 2018 06.
Article in English | MEDLINE | ID: mdl-29100480

ABSTRACT

Phenotypic cell-based assays have proven to be efficient at discovering first-in-class therapeutic drugs mainly because they allow for scanning a wide spectrum of possible targets at once. However, despite compelling methodological advances, posterior identification of a compound's mechanism of action (MOA) has remained difficult and highly refractory to automated analyses. Methods such as the cell painting assay and multiplexing fluorescent dyes to reveal broadly relevant cellular components were recently suggested for MOA prediction. We demonstrated that adding fluorescent dyes to a single assay has limited impact on MOA prediction accuracy, as monitoring only the nuclei stain could reach compelling levels of accuracy. This observation suggested that multiplexed measurements are highly correlated and nuclei stain could possibly reflect the general state of the cell. We then hypothesized that combining unrelated and possibly simple cell-based assays could bring a solution that would be biologically and technically more relevant to predict a drug target than using a single assay multiplexing dyes. We show that such a combination of past screen data could rationally be reused in screening facilities to train an ensemble classifier to predict drug targets and prioritize a possibly large list of unknown compound hits at once.


Subject(s)
Drug Evaluation, Preclinical/methods , Mesothelioma/drug therapy , Microscopy, Fluorescence/methods , Molecular Targeted Therapy/methods , Prostatic Neoplasms/drug therapy , Cell Line, Tumor , Datasets as Topic , Fluorescent Dyes/metabolism , Humans , Male , Mesothelioma/diagnosis , Mesothelioma/pathology , Phenotype , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/pathology , Staining and Labeling
3.
Nat Commun ; 8: 15554, 2017 05 31.
Article in English | MEDLINE | ID: mdl-28561033

ABSTRACT

Three-dimensional fluorescence microscopy followed by image processing is routinely used to study biological objects at various scales such as cells and tissue. However, maximum intensity projection, the most broadly used rendering tool, extracts a discontinuous layer of voxels, obliviously creating important artifacts and possibly misleading interpretation. Here we propose smooth manifold extraction, an algorithm that produces a continuous focused 2D extraction from a 3D volume, hence preserving local spatial relationships. We demonstrate the usefulness of our approach by applying it to various biological applications using confocal and wide-field microscopy 3D image stacks. We provide a parameter-free ImageJ/Fiji plugin that allows 2D visualization and interpretation of 3D image stacks with maximum accuracy.

4.
Eur Respir J ; 46(6): 1762-72, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26541534

ABSTRACT

The impact of host immunity on outcome in nonsmall cell lung cancer (NSCLC) is controversial. We examined the relationship between lymphoid infiltration patterns in NSCLC and prognosis.Tumour- and stroma-infiltrating CD3(+), CD8(+) and forkhead box P3 (Foxp3)(+) T-lymphocytes were identified using immunohistochemistry and a novel image analysis algorithm to assess total, cytotoxic and regulatory T-lymphocyte counts, respectively, in 196 NSCLC cases. The median cell count was selected as a cut-point to define patient subgroups and the ratio of the corresponding tumour islet:stroma (TI/S) counts was determined.There was a positive association between overall survival and increased CD8(+) TI/S ratio (hazard ratio (HR) for death 0.44, p<0.001) but an inverse relationship between Foxp3(+) TI/S ratio and overall survival (HR 4.86, p<0.001). Patients with high CD8(+) islet (HR 0.48, p<0.001) and Foxp3(+) stromal (HR 0.23, p<0.001) counts had better survival, whereas high CD3(+) and CD8(+) stromal counts and high Foxp3(+) islet infiltration conferred a worse survival (HR 1.55, 2.19 and 3.14, respectively). By multivariate analysis, a high CD8(+) TI/S ratio conferred an improved survival (HR 0.48, p=0.002) but a high Foxp3(+) TI/S ratio was associated with worse survival (HR 3.91, p<0.001).Microlocalisation of infiltrating T-lymphocytes is a powerful predictor of outcome in resected NSCLC.


Subject(s)
Adenocarcinoma/immunology , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Large Cell/immunology , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Squamous Cell/immunology , Lung Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenocarcinoma/surgery , Adult , Aged , Aged, 80 and over , Algorithms , CD8-Positive T-Lymphocytes/metabolism , Carcinoma, Large Cell/genetics , Carcinoma, Large Cell/pathology , Carcinoma, Large Cell/surgery , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/surgery , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/surgery , Cohort Studies , ErbB Receptors/genetics , Female , Forkhead Transcription Factors/metabolism , Humans , Image Processing, Computer-Assisted , Immunohistochemistry , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/surgery , Lymphocyte Count , Male , Middle Aged , Multivariate Analysis , Neoplasm Staging , Neoplasm, Residual , Pneumonectomy , Prognosis , Proportional Hazards Models , Proto-Oncogene Proteins p21(ras)/genetics , Retrospective Studies , T-Lymphocytes, Regulatory/metabolism , Tumor Burden
5.
PLoS One ; 8(5): e62070, 2013.
Article in English | MEDLINE | ID: mdl-23690928

ABSTRACT

AIMS: Immunohistochemistry is a routine practice in clinical cancer diagnostics and also an established technology for tissue-based research regarding biomarker discovery efforts. Tedious manual assessment of immunohistochemically stained tissue needs to be fully automated to take full advantage of the potential for high throughput analyses enabled by tissue microarrays and digital pathology. Such automated tools also need to be reproducible for different experimental conditions and biomarker targets. In this study we present a novel supervised melanoma specific pattern recognition approach that is fully automated and quantitative. METHODS AND RESULTS: Melanoma samples were immunostained for the melanocyte specific target, Melan-A. Images representing immunostained melanoma tissue were then digitally processed to segment regions of interest, highlighting Melan-A positive and negative areas. Color deconvolution was applied to each region of interest to separate the channel containing the immunohistochemistry signal from the hematoxylin counterstaining channel. A support vector machine melanoma classification model was learned from a discovery melanoma patient cohort (n = 264) and subsequently validated on an independent cohort of melanoma patient tissue sample images (n = 157). CONCLUSION: Here we propose a novel method that takes advantage of utilizing an immuhistochemical marker highlighting melanocytes to fully automate the learning of a general melanoma cell classification model. The presented method can be applied on any protein of interest and thus provides a tool for quantification of immunohistochemistry-based protein expression in melanoma.


Subject(s)
Immunohistochemistry/methods , Melanocytes/chemistry , Melanoma/diagnosis , Pattern Recognition, Automated/methods , Tissue Array Analysis/methods , Humans , Image Processing, Computer-Assisted , MART-1 Antigen , Surface Properties
6.
Proc Natl Acad Sci U S A ; 110(17): 6853-8, 2013 Apr 23.
Article in English | MEDLINE | ID: mdl-23569271

ABSTRACT

The transformation of normal cells to malignant, metastatic tumor cells is a multistep process caused by the sequential acquirement of genetic changes. To identify these changes, we compared the transcriptomes and levels and distribution of proteins in a four-stage cell model of isogenically matched normal, immortalized, transformed, and metastatic human cells, using deep transcriptome sequencing and immunofluorescence microscopy. The data show that ∼6% (n = 1,357) of the human protein-coding genes are differentially expressed across the stages in the model. Interestingly, the majority of these genes are down-regulated, linking malignant transformation to dedifferentiation. The up-regulated genes are mainly components that control cellular proliferation, whereas the down-regulated genes consist of proteins exposed on or secreted from the cell surface. As many of the identified gene products control basic cellular functions that are defective in cancers, the data provide candidates for follow-up studies to investigate their functional roles in tumor formation. When we further compared the expression levels of four of the identified proteins in clinical cancer cohorts, similar differences were observed between benign and cancer cells, as in the cell model. This shows that this comprehensive demonstration of the molecular changes underlying malignant transformation is a relevant model to study the process of tumor formation.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Gene Expression Regulation, Neoplastic/physiology , Models, Biological , Proteins/metabolism , Antigens, Polyomavirus Transforming/metabolism , Cell Line, Tumor , Fibroblasts/metabolism , Gene Expression Profiling , Genes, ras/physiology , Genetic Association Studies , High-Throughput Nucleotide Sequencing , Humans , Immunohistochemistry , Microscopy, Fluorescence , Principal Component Analysis , Telomerase/metabolism
7.
BMC Cancer ; 13: 175, 2013 Apr 02.
Article in English | MEDLINE | ID: mdl-23547718

ABSTRACT

BACKGROUND: Although omic-based discovery approaches can provide powerful tools for biomarker identification, several reservations have been raised regarding the clinical applicability of gene expression studies, such as their prohibitive cost. However, the limited availability of antibodies is a key barrier to the development of a lower cost alternative, namely a discrete collection of immunohistochemistry (IHC)-based biomarkers. The aim of this study was to use a systematic approach to generate and screen affinity-purified, mono-specific antibodies targeting progression-related biomarkers, with a view towards developing a clinically applicable IHC-based prognostic biomarker panel for breast cancer. METHODS: We examined both in-house and publicly available breast cancer DNA microarray datasets relating to invasion and metastasis, thus identifying a cohort of candidate progression-associated biomarkers. Of these, 18 antibodies were released for extended analysis. Validated antibodies were screened against a tissue microarray (TMA) constructed from a cohort of consecutive breast cancer cases (n = 512) to test the immunohistochemical surrogate signature. RESULTS: Antibody screening revealed 3 candidate prognostic markers: the cell cycle regulator, Anillin (ANLN); the mitogen-activated protein kinase, PDZ-Binding Kinase (PBK); and the estrogen response gene, PDZ-Domain Containing 1 (PDZK1). Increased expression of ANLN and PBK was associated with poor prognosis, whilst increased expression of PDZK1 was associated with good prognosis. A 3-marker signature comprised of high PBK, high ANLN and low PDZK1 expression was associated with decreased recurrence-free survival (p < 0.001) and breast cancer-specific survival (BCSS) (p < 0.001). This novel signature was associated with high tumour grade (p < 0.001), positive nodal status (p = 0.029), ER-negativity (p = 0.006), Her2-positivity (p = 0.036) and high Ki67 status (p < 0.001). However, multivariate Cox regression demonstrated that the signature was not a significant predictor of BCSS (HR = 6.38; 95% CI = 0.79-51.26, p = 0.082). CONCLUSIONS: We have developed a comprehensive biomarker pathway that extends from discovery through to validation on a TMA platform. This proof-of-concept study has resulted in the identification of a novel 3-protein prognostic panel. Additional biochemical markers, interrogated using this high-throughput platform, may further augment the prognostic accuracy of this panel to a point that may allow implementation into routine clinical practice.


Subject(s)
Antibodies, Monoclonal , Biomarkers, Tumor/analysis , Breast Neoplasms/metabolism , Carrier Proteins/biosynthesis , Contractile Proteins/biosynthesis , Mitogen-Activated Protein Kinase Kinases/biosynthesis , Adult , Aged , Aged, 80 and over , Blotting, Western , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Carrier Proteins/analysis , Contractile Proteins/analysis , Female , High-Throughput Screening Assays , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Membrane Proteins , Middle Aged , Mitogen-Activated Protein Kinase Kinases/analysis , Oligonucleotide Array Sequence Analysis , Prognosis , Proportional Hazards Models , Tissue Array Analysis
8.
Nat Methods ; 10(4): 315-23, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23435261

ABSTRACT

Imaging techniques such as immunofluorescence (IF) and the expression of fluorescent protein (FP) fusions are widely used to investigate the subcellular distribution of proteins. Here we report a systematic analysis of >500 human proteins comparing the localizations obtained in live versus fixed cells using FPs and IF, respectively. We identify systematic discrepancies between IF and FPs as well as between FP tagging at the N and C termini. The analysis shows that for 80% of the proteins, IF and FPs yield the same subcellular distribution, and the locations of 250 previously unlocalized proteins were determined by the overlap between the two methods. Approximately 60% of proteins localize to multiple organelles for both methods, indicating a complex subcellular protein organization. These results show that both IF and FP tagging are reliable techniques and demonstrate the usefulness of an integrative approach for a complete investigation of the subcellular human proteome.


Subject(s)
Fluorescent Antibody Technique , Luminescent Proteins/metabolism , Protein Transport/physiology , Staining and Labeling/methods , Animals , Chlorocebus aethiops , Gene Expression Regulation , HeLa Cells , Humans , Luminescent Proteins/genetics , Protein Interaction Mapping , Vero Cells
9.
Cancer Discov ; 1(1): 54-67, 2011 Jun.
Article in English | MEDLINE | ID: mdl-22039576

ABSTRACT

UNLABELLED: Immune-regulated pathways influence multiple aspects of cancer development. In this article we demonstrate that both macrophage abundance and T-cell abundance in breast cancer represent prognostic indicators for recurrence-free and overall survival. We provide evidence that response to chemotherapy is in part regulated by these leukocytes; cytotoxic therapies induce mammary epithelial cells to produce monocyte/macrophage recruitment factors, including colony stimulating factor 1 (CSF1) and interleukin-34, which together enhance CSF1 receptor (CSF1R)-dependent macrophage infiltration. Blockade of macrophage recruitment with CSF1R-signaling antagonists, in combination with paclitaxel, improved survival of mammary tumor-bearing mice by slowing primary tumor development and reducing pulmonary metastasis. These improved aspects of mammary carcinogenesis were accompanied by decreased vessel density and appearance of antitumor immune programs fostering tumor suppression in a CD8+ T-cell-dependent manner. These data provide a rationale for targeting macrophage recruitment/response pathways, notably CSF1R, in combination with cytotoxic therapy, and identification of a breast cancer population likely to benefit from this novel therapeutic approach. SIGNIFICANCE: These findings reveal that response to chemotherapy is in part regulated by the tumor immune microenvironment and that common cytotoxic drugs induce neoplastic cells to produce monocyte/macrophage recruitment factors, which in turn enhance macrophage infiltration into mammary adenocarcinomas. Blockade of pathways mediating macrophage recruitment, in combination with chemotherapy, significantly decreases primary tumor progression, reduces metastasis, and improves survival by CD8+ T-cell-dependent mechanisms, thus indicating that the immune microenvironment of tumors can be reprogrammed to instead foster antitumor immunity and improve response to cytotoxic therapy.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Leukocytes/immunology , Macrophages/immunology , Aged , Animals , Breast Neoplasms/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cohort Studies , Disease-Free Survival , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/metabolism , Female , Follow-Up Studies , Humans , Leukocytes/drug effects , Leukocytes/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Macrophage Colony-Stimulating Factor/immunology , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/metabolism , Mice , Neoplasm Metastasis , Paclitaxel/pharmacology , Prognosis , Receptor, Macrophage Colony-Stimulating Factor/immunology , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Survival Rate , Tumor Microenvironment/immunology
10.
BMC Pregnancy Childbirth ; 11: 74, 2011 Oct 16.
Article in English | MEDLINE | ID: mdl-21999197

ABSTRACT

BACKGROUND: The most common indication for cesarean section (CS) in nulliparous women is dystocia secondary to ineffective myometrial contractility. The aim of this study was to identify a molecular profile in myometrium associated with dystocic labor. METHODS: Myometrial biopsies were obtained from the upper incisional margins of nulliparous women undergoing lower segment CS for dystocia (n = 4) and control women undergoing CS in the second stage who had demonstrated efficient uterine action during the first stage of labor (n = 4). All patients were in spontaneous (non-induced) labor and had received intrapartum oxytocin to accelerate labor. RNA was extracted from biopsies and hybridized to Affymetrix HuGene U133A Plus 2 microarrays. Internal validation was performed using quantitative SYBR Green Real-Time PCR. RESULTS: Seventy genes were differentially expressed between the two groups. 58 genes were down-regulated in the dystocia group. Gene ontology analysis revealed 12 of the 58 down-regulated genes were involved in the immune response. These included (ERAP2, (8.67 fold change (FC)) HLA-DQB1 (7.88 FC) CD28 (2.60 FC), LILRA3 (2.87 FC) and TGFBR3 (2.1 FC)) Hierarchical clustering demonstrated a difference in global gene expression patterns between the samples from dystocic and non-dystocic labours. RT-PCR validation was performed on 4 genes ERAP2, CD28, LILRA3 and TGFBR3 CONCLUSION: These findings suggest an underlying molecular basis for dystocia in nulliparous women in spontaneous labor. Differentially expressed genes suggest an important role for the immune response in dystocic labor and may provide important indicators for new diagnostic assays and potential intrapartum therapeutic targets.


Subject(s)
Dystocia/diagnosis , Myometrium/metabolism , Uterine Contraction , Adult , Aminopeptidases/genetics , Case-Control Studies , Cesarean Section , Down-Regulation , Dystocia/genetics , Female , Humans , Parity , Polymerase Chain Reaction , Pregnancy , RNA/analysis
11.
BMC Cancer ; 11: 405, 2011 Sep 24.
Article in English | MEDLINE | ID: mdl-21943380

ABSTRACT

BACKGROUND: The neural transcription factor SOX11 has been described as a prognostic marker in epithelial ovarian cancers (EOC), however its role in individual histological subtypes and tumour grade requires further clarification. Furthermore, methylation-dependent silencing of SOX11 has been reported for B cell lymphomas and indicates that epigenetic drugs may be used to re-express this tumour suppressor, but information on SOX11 promoter methylation in EOC is still lacking. METHODS: SOX11 expression and clinicopathological data was compared using χ² test in a cohort of 154 cases of primary invasive EOC. Kaplan-Meier analysis and the log rank test were applied to evaluate ovarian cancer-specific survival (OCSS) and overall survival (OS) in strata, according to SOX11 expression. Also, the methylation status of the SOX11 promoter was determined by sodium bisulfite sequencing and methylation specific PCR (MSP). Furthermore, the effect of ectopic overexpression of SOX11 on proliferation was studied through [3H]-thymidine incorporation. RESULTS: SOX11 expression was associated with an improved survival of patients with high grade EOC, although not independent of stage. Further analyses of EOC cell lines showed that SOX11 mRNA and protein were expressed in two of five cell lines, correlating with promoter methylation status. Demethylation was successfully performed using 5'-Aza-2'deoxycytidine (5-Aza-dC) resulting in SOX11 mRNA and protein expression in a previously negative EOC cell line. Furthermore, overexpression of SOX11 in EOC cell lines confirmed the growth regulatory role of SOX11. CONCLUSIONS: SOX11 is a functionally associated protein in EOC with prognostic value for high-grade tumours. Re-expression of SOX11 in EOC indicates a potential use of epigenetic drugs to affect cellular growth in SOX11-negative tumours.


Subject(s)
DNA Methylation , Neoplasms, Glandular and Epithelial/genetics , Neoplasms, Glandular and Epithelial/mortality , Ovarian Neoplasms/genetics , Ovarian Neoplasms/mortality , Promoter Regions, Genetic , SOXC Transcription Factors/genetics , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Endometrial Neoplasms/genetics , Endometrial Neoplasms/mortality , Endometrial Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Staging , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/pathology , RNA, Messenger , SOXC Transcription Factors/metabolism
12.
Transl Oncol ; 4(4): 212-21, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21804916

ABSTRACT

The RNA-binding motif protein 3 (RBM3) was initially discovered as a putative cancer biomarker based on its differential expression in various cancer forms in the Human Protein Atlas (HPA). We previously reported an association between high expression of RBM3 and prolonged survival in breast and epithelial ovarian cancer (EOC). Because the function of RBM3 has not been fully elucidated, the aim of this study was to use gene set enrichment analysis to identify the underlying biologic processes associated with RBM3 expression in a previously analyzed EOC cohort (cohort 1, n = 267). This revealed an association between RBM3 expression and several cellular processes involved in the maintenance of DNA integrity. RBM3-regulated genes were subsequently screened in the HPA to select for putative prognostic markers, and candidate proteins were analyzed in the ovarian cancer cell line A2780, whereby an up-regulation of Chk1, Chk2, and MCM3 was demonstrated in siRBM3-treated cells compared to controls. The prognostic value of these markers was assessed at the messenger RNA level in cohort 1 and the protein level in an independent EOC cohort (cohort 2, n = 154). High expression levels of Chk1, Chk2, and MCM3 were associated with a significantly shorter survival in both cohorts, and phosphorylated Chk2 was an adverse prognostic marker in cohort 2. These results uncover a putative role for RBM3 in DNA damage response, which might, in part, explain its cisplatin-sensitizing properties and good prognostic value in EOC. Furthermore, it is demonstrated that Chk1, Chk2, and MCM3 are poor prognostic markers in EOC.

13.
Clin Cancer Res ; 17(18): 6083-96, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21795409

ABSTRACT

PURPOSE: We investigated whether inhibition of interleukin 6 (IL-6) has therapeutic activity in ovarian cancer via abrogation of a tumor-promoting cytokine network. EXPERIMENTAL DESIGN: We combined preclinical and in silico experiments with a phase 2 clinical trial of the anti-IL-6 antibody siltuximab in patients with platinum-resistant ovarian cancer. RESULTS: Automated immunohistochemistry on tissue microarrays from 221 ovarian cancer cases showed that intensity of IL-6 staining in malignant cells significantly associated with poor prognosis. Treatment of ovarian cancer cells with siltuximab reduced constitutive cytokine and chemokine production and also inhibited IL-6 signaling, tumor growth, the tumor-associated macrophage infiltrate and angiogenesis in IL-6-producing intraperitoneal ovarian cancer xenografts. In the clinical trial, the primary endpoint was response rate as assessed by combined RECIST and CA125 criteria. One patient of eighteen evaluable had a partial response, while seven others had periods of disease stabilization. In patients treated for 6 months, there was a significant decline in plasma levels of IL-6-regulated CCL2, CXCL12, and VEGF. Gene expression levels of factors that were reduced by siltuximab treatment in the patients significantly correlated with high IL-6 pathway gene expression and macrophage markers in microarray analyses of ovarian cancer biopsies. CONCLUSION: IL-6 stimulates inflammatory cytokine production, tumor angiogenesis, and the tumor macrophage infiltrate in ovarian cancer and these actions can be inhibited by a neutralizing anti-IL-6 antibody in preclinical and clinical studies.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Interleukin-6/antagonists & inhibitors , Ovarian Neoplasms/drug therapy , Animals , Antibodies, Monoclonal/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Biomarkers/blood , Cell Line, Tumor , Computational Biology , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Interleukin-6/metabolism , Kaplan-Meier Estimate , Mice , Mice, Inbred BALB C , Mice, Nude , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/mortality , Prognosis , Receptors, Interleukin-6/metabolism , Signal Transduction , Treatment Outcome , Xenograft Model Antitumor Assays
14.
Am J Surg Pathol ; 35(7): 937-48, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21677534

ABSTRACT

The special AT-rich sequence-binding protein 2 (SATB2), a nuclear matrix-associated transcription factor and epigenetic regulator, was identified as a tissue type-specific protein when screening protein expression patterns in human normal and cancer tissues using an antibody-based proteomics approach. In this respect, the SATB2 protein shows a selective pattern of expression and, within cells of epithelial lineages, SATB2 expression is restricted to glandular cells lining the lower gastrointestinal tract. The expression of SATB2 protein is primarily preserved in cancer cells of colorectal origin, indicating that SATB2 could function as a clinically useful diagnostic marker to distinguish colorectal cancer (CRC) from other types of cancer. The aim of this study was to further explore and validate the specific expression pattern of SATB2 as a clinical biomarker and to compare SATB2 with the well-known cytokeratin 20 (CK20). Immunohistochemistry was used to analyze the extent of SATB2 expression in tissue microarrays with tumors from 9 independent cohorts of patients with primary and metastatic CRCs (n=1882). Our results show that SATB2 is a sensitive and highly specific marker for CRC with distinct positivity in 85% of all CRCs, and that SATB2 and/or CK20 was positive in 97% of CRCs. In conclusion, the specific expression of SATB2 in a large majority of CRCs suggests that SATB2 can be used as an important complementary tool for the differential diagnosis of carcinoma of unknown primary origin.


Subject(s)
Adenocarcinoma/diagnosis , Colorectal Neoplasms/diagnosis , Keratin-20/metabolism , Matrix Attachment Region Binding Proteins/metabolism , Transcription Factors/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/secondary , Adenoma , Biomarkers, Tumor/metabolism , Cohort Studies , Colorectal Neoplasms/metabolism , Diagnosis, Differential , Female , Humans , Image Processing, Computer-Assisted , Immunohistochemistry/methods , Male , Neoplasm Staging , Predictive Value of Tests , Tissue Array Analysis
15.
Mod Pathol ; 24(5): 708-19, 2011 May.
Article in English | MEDLINE | ID: mdl-21240253

ABSTRACT

Despite prostate cancer being the most frequent cancer in men in the Western world, tissue biomarkers for predicting disease recurrence after surgery have not been incorporated into clinical practice. Our group has previously identified ß-microseminoprotein (MSMB) and cysteine-rich secretory protein-3 (CRISP3) as independent predictors of biochemical recurrence after radical prostatectomy. The purpose of the present study was to use automated image analysis, enabling quantitative determination of MSMB and CRISP3 expressions in a large cohort and to validate the previous findings. MSMB and CRISP3 protein expressions were assessed on tissue microarrays constructed from 3268 radical prostatectomy specimens. Whole-slide digital images were captured, and a novel cytoplasmic algorithm was used to develop a quantitative scoring model for cytoplasmic staining. Classification regression tree analysis was used to group patients, with different risk for biochemical recurrence, depending on level of protein expression. Patients with tumors expressing high levels of MSMB had a significantly reduced risk for biochemical recurrence after radical prostatectomy (HR=0.468; 95% CI 0.394-0.556; P<0.001). Multivariate analysis adjusted for clinicopathological parameters revealed that MSMB expression was an independent predictor of decreased risk of recurrence (HR=0.710; 95% CI 0.578-0.872; P<0.001). We found no correlation between CRISP3 expression and biochemical recurrence. In this current study, we applied a novel image analysis on a large independent cohort and successfully verified that MSMB is a strong independent factor, predicting favorable outcome after radical prostatectomy for localized prostate cancer.


Subject(s)
Adenocarcinoma/secondary , Image Processing, Computer-Assisted/methods , Prostatic Neoplasms/pathology , Prostatic Secretory Proteins/metabolism , Salivary Proteins and Peptides/metabolism , Seminal Plasma Proteins/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Aged , Algorithms , Biomarkers, Tumor/metabolism , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Recurrence, Local , Prostatectomy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/mortality , Survival Rate , Sweden/epidemiology , Tissue Array Analysis
16.
Methods Mol Biol ; 691: 97-112, 2011.
Article in English | MEDLINE | ID: mdl-20972749

ABSTRACT

Tissue microarrays (TMAs) have recently emerged as very valuable tools for high-throughput pathological assessment, especially in the cancer research arena. This important technology, however, has yet to fully penetrate into the area of toxicology. Here, we describe the creation of TMAs representative of samples produced from conventional toxicology studies within a large-scale, multi-institutional pan-European project, PredTox. PredTox, short for Predictive Toxicology, formed part of an EU FP6 Integrated Project, Innovative Medicines for Europe (InnoMed), and aimed to study pre-clinically 16 compounds of known liver and/or kidney toxicity. In more detail, TMAs were constructed from materials corresponding to the full face sections of liver and kidney from rats treated with different drug candidates by members of the consortium. We also describe the process of digital slide scanning of kidney and liver sections, in the context of creating an online resource of histopathological data.


Subject(s)
Image Processing, Computer-Assisted/methods , Tissue Array Analysis/methods , Animals , Frozen Sections , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Information Storage and Retrieval , Rats
17.
BMC Cancer ; 10: 639, 2010 Nov 23.
Article in English | MEDLINE | ID: mdl-21092276

ABSTRACT

BACKGROUND: Conflicting data exist regarding the prognostic and predictive impact of survivin (BIRC5) in breast cancer. We previously reported survivin cytoplasmic-to-nuclear ratio (CNR) as an independent prognostic indicator in breast cancer. Here, we validate survivin CNR in a separate and extended cohort. Furthermore, we present new data suggesting that a low CNR may predict outcome in tamoxifen-treated patients. METHODS: Survin expression was assessed using immunhistochemistry on a breast cancer tissue microarray (TMA) containing 512 tumours. Whole slide digital images were captured using an Aperio XT scanner. Automated image analysis was used to identify tumour from stroma and then to quantify tumour-specific nuclear and cytoplasmic survivin. A decision tree model selected using a 10-fold cross-validation approach was used to identify prognostic subgroups based on nuclear and cytoplasmic survivin expression. RESULTS: Following optimisation of the staining procedure, it was possible to evaluate survivin protein expression in 70.1% (n = 359) of the 512 tumours represented on the TMA. Decision tree analysis predicted that nuclear, as opposed to cytoplasmic, survivin was the most important determinant of overall survival (OS) and breast cancer-specific survival (BCSS). The decision tree model confirmed CNR of 5 as the optimum threshold for survival analysis. Univariate analysis demonstrated an association between a high CNR (>5) and a prolonged BCSS (HR 0.49, 95% CI 0.29-0.81, p = 0.006). Multivariate analysis revealed a high CNR (>5) was an independent predictor of BCSS (HR 0.47, 95% CI 0.27-0.82, p = 0.008). An increased CNR was associated with ER positive (p = 0.045), low grade (p = 0.007), Ki-67 (p = 0.001) and Her2 (p = 0.026) negative tumours. Finally, a high CNR was an independent predictor of OS in tamoxifen-treated ER-positive patients (HR 0.44, 95% CI 0.23-0.87, p = 0.018). CONCLUSION: Using the same threshold as our previous study, we have validated survivin CNR as a marker of good prognosis in breast cancer in a large independent cohort. These findings provide robust evidence of the importance of survivin CNR as a breast cancer biomarker, and its potential to predict outcome in tamoxifen-treated patients.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/chemistry , Cell Nucleus/chemistry , Cytoplasm/chemistry , Microtubule-Associated Proteins/analysis , Adult , Aged , Aged, 80 and over , Algorithms , Breast Neoplasms/mortality , Breast Neoplasms/therapy , Chi-Square Distribution , Decision Trees , Estrogen Receptor Modulators/therapeutic use , Female , Humans , Image Interpretation, Computer-Assisted , Immunohistochemistry , Inhibitor of Apoptosis Proteins , Kaplan-Meier Estimate , Ki-67 Antigen/analysis , Middle Aged , Pattern Recognition, Automated , Predictive Value of Tests , Proportional Hazards Models , Receptor, ErbB-2/analysis , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , Reproducibility of Results , Risk Assessment , Risk Factors , Survivin , Sweden , Tamoxifen/therapeutic use , Time Factors , Tissue Array Analysis , Treatment Outcome
18.
Cancer Res ; 70(20): 7776-87, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20924104

ABSTRACT

Using transgenic mouse models of breast cancer that ablate Src homology and collagen A (ShcA) expression or oncogene-coupled ShcA signaling, we previously showed that this adaptor is critical for mammary tumor onset and progression. We now provide the first evidence that ShcA regulates mammary tumorigenesis, in part, through its ability to regulate the adaptive immune response. Inactivation of ShcA signaling within tumor cells results in extensive CD4(+) T-cell infiltration and induction of a humoral immune response in mammary tumors. This is associated with a robust CTL response in preneoplastic lesions that are deficient in ShcA signaling. Moreover, mammary tumor progression of ShcA-deficient hyperplasias is accelerated in a T cell-deficient background. We also uncover a clinically relevant correlation between high ShcA expression and low CTL infiltration in human breast cancers. Finally, we define a novel ShcA-regulated immune signature that functions as an independent prognostic marker of survival in human epidermal growth factor receptor 2(+) and basal breast cancers. We reveal a novel role for tumor cell-derived ShcA in the establishment and maintenance of an immunosuppressive state.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Receptor Protein-Tyrosine Kinases/physiology , Shc Signaling Adaptor Proteins/genetics , Animals , Breast Neoplasms/immunology , CD4-Positive T-Lymphocytes/pathology , Female , Humans , Immunosuppression Therapy , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Parity , Pregnancy , Proportional Hazards Models , RNA, Neoplasm/genetics , RNA, Neoplasm/isolation & purification , Shc Signaling Adaptor Proteins/deficiency , Shc Signaling Adaptor Proteins/physiology , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Treatment Outcome
19.
Melanoma Res ; 20(6): 468-78, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20890226

ABSTRACT

SOX10 is a transcription factor expressed in nerve cells and melanocytes. The aim of this study was to investigate the protein expression pattern of SOX10 in malignant melanoma tumors and to analyze whether the results correlated with clinical parameters and the proliferation marker Ki-67. Furthermore, proliferation and migration were analyzed in three different cell lines employing SOX10 small interfering RNA-mediated silencing. Expression patterns were determined in 106 primary tumors and 39 metastases in addition to 16 normal skin samples and six benign nevi employing immunohistochemistry and tissue microarrays. The immunohistochemical staining was evaluated manually and with an automated algorithm. SOX10 was strongly expressed in the benign tissues, but for the malignant tumors superficial spreading melanomas stained stronger than nodular malignant melanomas (P=0.008). The staining intensity was also inversely correlated with T-stage (Spearman's ρ=-0.261, P=0.008). Overall survival and time to recurrence were significantly correlated with SOX10 intensity, but not in multivariate analysis including T-stage. With the automated algorithm there was an inverse correlation between the SOX10 staining intensity and the proliferation marker, Ki-67 (ρ=-0.173, P=0.02) and a significant difference in the intensity signal between the benign tissues, the primary tumors and the metastases where the metastases stained the weakest (P≤0.001). SOX10 downregulation resulted in variable effects on proliferation and migration rates in the melanoma cell lines. In conclusion, the SOX10 intensity level differed depending on the tissue studied and SOX10 might have a role in survival. No conclusion regarding the role of SOX10 for in-vitro proliferation and migration could be drawn.


Subject(s)
Melanoma/metabolism , SOXE Transcription Factors/biosynthesis , Skin Neoplasms/metabolism , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Movement/physiology , Down-Regulation , Humans , Immunohistochemistry , Melanoma/genetics , Melanoma/pathology , Neoplasm Metastasis , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , SOXE Transcription Factors/genetics , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Transfection
20.
Nat Rev Cancer ; 10(9): 605-17, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20720569

ABSTRACT

The effective implementation of personalized cancer therapeutic regimens depends on the successful identification and translation of informative biomarkers to aid clinical decision making. Antibody-based proteomics occupies a pivotal space in the cancer biomarker discovery and validation pipeline, facilitating the high-throughput evaluation of candidate markers. Although the clinical utility of these emerging technologies remains to be established, the traditional use of antibodies as affinity reagents in clinical diagnostic and predictive assays suggests that the rapid translation of such approaches is an achievable goal. Furthermore, in combination with, or as alternatives to, genomic and transcriptomic methods for patient stratification, antibody-based proteomics approaches offer the promise of additional insight into cancer disease states. In this Review, we discuss the current status of antibody-based proteomics and its contribution to the development of new assays that are crucial for the realization of individualized cancer therapy.


Subject(s)
Antibodies/immunology , Biomarkers, Tumor/analysis , Neoplasms/diagnosis , Proteomics/methods , Biomarkers, Tumor/immunology , Humans , Neoplasms/chemistry , Neoplasms/immunology , Neoplasms/therapy , Prognosis
SELECTION OF CITATIONS
SEARCH DETAIL
...